throbber
Guidance for Industry
`
`Clinical Trial Endpoints
`
`for the Approval of Cancer
`
`Drugs and Biologics
`
`
`
`
`
`
`
`U.S. Department of Health and Human Services
`
`Food and Drug Administration
`
`Center for Drug Evaluation and Research (CDER)
`
`Center for Biologics Evaluation and Research (CBER)
`
`
`May 2007
`
`Clinical/Medical
`
`
`
`
`
`
`NPC02232922
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 1 of 22
`
`

`
`Guidance for Industry
`
`Clinical Trial Endpoints
`
`for the Approval of Cancer
`
`Drugs and Biologics
`
`
`
`Additional copies are available from:
`
`
`
`
`
`Office of Training and Communications
`
`
`
`Division of Drug Information, HFD-240
`
`
`
`Center for Drug Evaluation and Research
`
`
`Food and Drug Administration
`
`5600 Fishers Lane
`
`
`Rockville, MD 20857
`
`
`(Tel) 301-827-4573
`
`http://www.fda.gov/cder/guidance/index.htm
`
`
`
`
`or
`
`
`
`Office of Communication, Training, and
`
`Manufacturers Assistance, HFM-40
`
`Center for Biologics Evaluation and Research
`
`
`Food and Drug Administration
`
`
`1401 Rockville Pike, Rockville, MD 20852-1448
`
`
`
`(Tel) 800-835-4709 or 301-827-1800
`
`
`http://www.fda.gov/cber/guidelines.htm
`
`
`
`
`U.S. Department of Health and Human Services
`
`Food and Drug Administration
`
`Center for Drug Evaluation and Research (CDER)
`
`Center for Biologics Evaluation and Research (CBER)
`
`
`May 2007
`
`Clinical/Medical
`
`
`
`
`
`
`NPC02232923
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 2 of 22
`
`

`
`
`
`TABLE OF CONTENTS
`
`
`
` I.
`INTRODUCTION............................................................................................................. 1
`
`
`
`BACKGROUND ............................................................................................................... 2
`
`II.
`
`
`
`
`A. Regulatory Requirements for Effectiveness ................................................................................2
`
`
`
`B. Endpoints Supporting Past Approvals in Oncology ...................................................................3
`
`III. GENERAL ENDPOINT CONSIDERATIONS ............................................................. 4
`
`
`
`
`
`A. Overall Survival .............................................................................................................................5
`
`
`
`B. Endpoints Based on Tumor Assessments.....................................................................................6
`
`
`
`1. Disease-Free Survival......................................................................................................................6
`
`
`
`2. Objective Response Rate..................................................................................................................7
`
`
`
`3. Time to Progression and Progression-Free Survival ......................................................................8
`
`
`
`a. TTP vs. PFS..............................................................................................................................8
`
`
`
`b. PFS as an endpoint to support drug approval ...........................................................................8
`
`
`
`c. PFS trial design issues ..............................................................................................................8
`
`
`
`d. Analysis of PFS ........................................................................................................................9
`
`
`
`4. Time-to-Treatment Failure ..............................................................................................................9
`
`
`
`C. Endpoints Involving Symptom Assessment...............................................................................10
`
`
`
`1. Specific Symptom Endpoints..........................................................................................................10
`
`
`
`2. Problems Encountered with Symptom Data ..................................................................................10
`
`
`
`D. Biomarkers ...................................................................................................................................11
`
`IV. CLINICAL TRIAL DESIGN CONSIDERATIONS ................................................... 11
`
`
`
`
`
`A. Single-Arm Studies ......................................................................................................................11
`
`
`
`B. Studies Designed to Demonstrate Noninferiority......................................................................11
`
`
`
`C. Trial Designs for Radiotherapy Protectants and Chemotherapy Protectants .......................12
`
`CONCLUSION ............................................................................................................... 12
`
`V.
`
`
`APPENDIX 1: TUMOR MEASUREMENT DATA COLLECTION.................................... 14
`
`
`
`APPENDIX 2: ISSUES TO CONSIDER IN PFS ANALYSIS ............................................... 15
`
`
`
`APPENDIX 3: EXAMPLE TABLES FOR PFS ANALYSIS................................................. 17
`
`
`
`APPENDIX 4: INDEPENDENT REVIEW OF TUMOR ENDPOINTS............................... 19
`
`
`
`
`
`NPC02232924
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 3 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`Guidance for Industry1
`
`Clinical Trial Endpoints for the Approval
`
`of Cancer Drugs and Biologics
`
`
`
`
`
`
`This guidance represents the Food and Drug Administration’s (FDA’s) current thinking on this topic. It
`does not create or confer any rights for or on any person and does not operate to bind FDA or the public.
`
`You can use an alternative approach if the approach satisfies the requirements of the applicable statutes
`and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for
`implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate
`number listed on the title page of this guidance.
`
`
`
`INTRODUCTION
`
`
`
`I.
`
`This guidance is the first in a planned series of cancer endpoint guidances. It provides
`recommendations to applicants on endpoints for cancer clinical trials submitted to the Food and
`Drug Administration (FDA) to support effectiveness claims in new drug applications (NDAs),
`biologics license applications (BLAs), or supplemental applications.2 It also provides
`background information and discusses general regulatory principles. The endpoints discussed in
`this guidance are for drugs to treat patients with an existing cancer. This guidance does not
`address endpoints for drugs to prevent or decrease the incidence of cancer.
`
`
`The FDA is developing guidance on oncology endpoints through a process that includes public
`workshops and discussions before the FDA’s Oncologic Drugs Advisory Committee (ODAC).3
`
`Each subsequent guidance will focus on endpoints for specific cancer types (e.g., lung cancer,
`colon cancer) to support drug approval or labeling claims.
`
`FDA’s guidance documents, including this guidance, do not establish legally enforceable
`responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should
`be viewed only as recommendations, unless specific regulatory or statutory requirements are
`cited. The use of the word should in Agency guidances means that something is suggested or
`recommended, but not required.
`
`
`
` 1 This guidance has been prepared by the Division of Drug Oncology Products and the Division of Biologic
`
`
`
` Oncology Products in the Center for Drug Evaluation and Research (CDER) in cooperation with the Center for
`
` Biologics Evaluation and Research (CBER) at the Food and Drug Administration.
`
`
`2 For the purposes of this guidance, all references to drugs include both human drugs and biological products unless
`
`
`otherwise specified.
`
`
`3 Transcripts are available at http://www.fda.gov/cder/drug/cancer_endpoints/default.htm.
`
`
`
`
`1
`
`
`NPC02232925
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 4 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`BACKGROUND
`
`
`
`II.
`
`Clinical trial endpoints serve different purposes. In conventional oncology drug development,
`early phase clinical trials evaluate safety and identify evidence of biological drug activity, such
`as tumor shrinkage. Endpoints for later phase efficacy studies commonly evaluate whether a
`drug provides a clinical benefit such as prolongation of survival or an improvement in
`symptoms. The following sections discuss the general regulatory requirements for efficacy and
`how they have influenced endpoint selection for the approval of cancer drugs. Later sections
`describe these endpoints in more detail and discuss whether they might serve as measures of
`disease activity or clinical benefit in various clinical settings.
`
`
`
`A.
`
`Regulatory Requirements for Effectiveness
`
`The requirement that new drugs show effectiveness is based on a 1962 amendment to the Federal
`Food, Drug, and Cosmetic Act. This law requires substantial evidence of effectiveness and
`specifies that this evidence must be derived from adequate and well-controlled clinical
`
`investigations. Similarly, the Public Health Service Act requires biological products to be safe,
`pure, and potent. Clinical benefits that have supported drug approval have included important
`clinical outcomes (e.g., increased survival, symptomatic improvement) but have also included
`effects on established surrogate endpoints (e.g., blood pressure, serum cholesterol).
`
`The accelerated approval regulations (21 CFR part 314, subpart H and 21 CFR part 601, subpart
`E), promulgated in 1992, allow use of additional endpoints for approval of drugs or biological
`products that are intended to treat serious or life-threatening diseases and that either demonstrate
`an improvement over available therapy or provide therapy where none exists. In this setting, the
`FDA may grant approval based on an effect on a surrogate endpoint that is reasonably likely to
`predict clinical benefit (“based on epidemiologic, therapeutic, pathophysiologic, or other
`evidence”). Such surrogates are less well-established than surrogates in regular use, such as
`blood pressure or cholesterol for cardiovascular disease. A drug is approved under the
`accelerated approval regulations on condition that the manufacturer conducts clinical studies to
`verify and describe the actual clinical benefit. If the postmarketing studies fail to demonstrate
`clinical benefit or if the applicant does not demonstrate due diligence in conducting the required
`studies, the drug may be removed from the market under an expedited process. In the following
`discussion, the term regular approval denotes the longstanding route of drug approval based on
`
`the demonstration of clinical benefit. That term is distinguished from accelerated approval,
`
`which is associated with use of a surrogate endpoint that is reasonably likely to predict benefit.
`
`The evidence critical for supporting drug approval, including the preferred number of clinical
`trials, is discussed in the guidance for industry FDA Approval of New Cancer Treatment Uses for
`Marketed Drug and Biological Products4 and in the FDA Modernization Act of 1997.5 In most
`
`
`
` 4 We update guidances periodically. To make sure you have the most recent version of a guidance, check the CDER
`
`
` guidance Web page at http://www.fda.gov/cder/guidance/index.htm.
`
`
`
`5 http://www.fda.gov/cder/fdama/default.htm
`
`
`
`
`
`2
`
`
`NPC02232926
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 5 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`
`cases, the FDA recommends at least two adequate and well-controlled clinical trials. In certain
`cases, evidence from a single trial can be sufficient (e.g., in cases in which a single multicenter
`study provides highly reliable and statistically strong evidence of an important clinical benefit,
`such as an effect on survival, and in which confirmation of the result in a second trial would be
`practically or ethically impossible). For drugs approved for treatment of patients with a specific
`stage of a particular malignancy, evidence from one trial may be sufficient to support an efficacy
`supplement for treatment of a different stage of the same cancer.
`
`
`
`B.
`
`Endpoints Supporting Past Approvals in Oncology
`
`For regular approval, it is critical that the applicant show direct evidence of clinical benefit or
`improvement in an established surrogate for clinical benefit. In oncology, survival improvement
`is considered an appropriate measure of clinical benefit. In addition, sponsors have used other
`endpoints for cancer drug approval. In the 1970s, the FDA usually approved cancer drugs based
`on objective response rate (ORR), determined by tumor assessments from radiological tests or
`physical examinations. In the early 1980s, after discussion with the ODAC, the FDA determined
`
`that cancer drug approval should be based on more direct evidence of clinical benefit, such as
`improvement in survival, improvement in a patient’s quality of life (QOL), improved physical
`functioning, or improved tumor-related symptoms. These benefits may not always be predicted
`by, or correlate with, ORR.
`
`Over the next decade, several endpoints were used as established surrogates for clinical benefit.
`Improvement in disease-free survival (DFS) supported drug approval in selected adjuvant
`settings, in which a large proportion of patients were expected to have cancer symptoms at the
`time of recurrence. Durable complete response was considered an established endpoint of
`clinical benefit in leukemia, where complete response is associated with less infection, bleeding,
`and blood product support. A high, substantiated ORR can support regular approval in select
`solid tumors, but that response duration, relief of tumor-related symptoms, and drug toxicity also
`should be considered when making the approval decision (O’Shaughnessy and Wittes et al.,
`1991, Commentary Concerning Demonstration of Safety and Efficacy of Investigational
`Anticancer Agents in Clinical Trials, J Clin Oncol, 9:2225-2232). For example, randomized
`trials for hormonal drugs for breast cancer have used ORR as an endpoint supporting regular
`approval. Improvement in tumor-related symptoms in conjunction with an improved ORR and
`adequate response duration has supported regular approval in several clinical settings.
`
`Surrogate endpoints for accelerated approval must be reasonably likely to predict clinical benefit
`(21 CFR part 314, subpart H and 21 CFR part 601, subpart E). Such drugs also must provide a
`benefit over available therapy (21 CFR part 314, subpart H and 21 CFR part 601, subpart E).6
`
`ORR has been the most commonly used surrogate endpoint in support of accelerated approval.
`Tumor response is widely accepted by oncologists in guiding cancer treatments. Because ORR
`is directly attributable to drug effect, single-arm trials conducted in patients with refractory
`tumors where no available therapy exists provide an accurate assessment of ORR.
`
`
`6 See the guidance for industry Available Therapy (http://www.fda.gov/cder/guidance/index.htm).
`
`
`
`3
`
`
`NPC02232927
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 6 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`
`
`III. GENERAL ENDPOINT CONSIDERATIONS
`
`
`This section provides an overview of general issues in cancer drug development. A discussion of
`
`commonly used cancer endpoints is followed by a discussion of pertinent issues in cancer
`
`clinical trial design using these endpoints (future guidances will discuss specific treatment
`
`indication endpoints). The endpoints that are discussed in this section include overall survival,
`
`endpoints based on tumor assessments (e.g., DFS, ORR, complete response, time to progression
`
`(TTP), progression-free survival (PFS)), and endpoints based on symptom assessment. Table 1
`
`provides a comparison of endpoints in cancer drug approval. Many issues relating to the proper
`
`analysis of efficacy endpoints are addressed in the ICH guidance for industry E9 Statistical
`
`Principles for Clinical Trials.
`
`
`
`Table 1. A Comparison of Important Cancer Approval Endpoints
`
`Endpoint
` Regulatory
`Study Design
`Advantages
`Evidence
`
`Disadvantages
`
`
`
`• May involve larger studies
`• May be affected by
`
`crossover therapy and
`sequential therapy
`
`• Includes noncancer deaths
`
`
`• Blinding is often difficult
`
`
`• Data are frequently missing
`
`or incomplete
`
`• Clinical significance of
`small changes is unknown
`
`• Multiple analyses
`• Lack of validated
`
`instruments
`
`• Not statistically validated as
`surrogate for survival in all
`
`settings
`
`• Not precisely measured;
`subject to assessment bias,
`
`particularly in open-label
`studies
`
`
`
`• Definitions vary among
`studies
`
`
`
`continued
`
`
`• Universally
`accepted direct
`
`measure of benefit
`
`
`
`• Easily measured
`
`
`• Precisely
`measured
`
`
`
`• Patient perspective
`of direct clinical
`
`benefit
`
`
`
`• Randomized
`studies essential
`• Blinding not
`
`essential
`
`
`
`Overall
`Survival
`
`Clinical benefit
`for regular
`approval
`
`
`Clinical benefit
`for regular
`
`approval
`
`
`• Randomized
`
`
`ibl nde ud st id es
`
`
`
`
`
`• Smaller sample
`size and shorter
`
`follow-up
`necessary
`compared with
`survival studies
`
`
`
`
`
`• Randomized
`studies essential
`• Blinding preferred
`
`• Blinded review
`recommended
`
`
`Surrogate for
`accelerated
`
`approval or
`regular
`
`approval*
`
`4
`
`
`Symptom
`Endpoints
`(patient-
`
`reported
`outcomes)
`
`Disease-Free
`Survival
`
`
`
`
`
`NPC02232928
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 7 of 22
`
`

`
`
`Table 1, continued
`Endpoint
` Regulatory
`
`Evidence
`
`Contains Nonbinding Recommendations
`
`
`
` Study Design
`
`Advantages
`
`Disadvantages
`
`
`
`
`• Not statistically validated as
`surrogate for survival in all
`settings
`
`
`• Not precisely measured;
`subject to assessment bias
`particularly in open-label
`
`studies
`
`• Definitions vary among
`
`
`studies
`
`• Frequent radiological or
`
`
`other assessments
`
`• Involves balanced timing of
`
`assessments among
`treatment arms
`
`
`
`
`
`
`
`• Single-arm or
`randomized
`
`studies can be
`used
`
`
`• Blinding
`preferred in
`
`comparative
`studies
`
`• Blinded review
`
`recommended
`
`
`• Single-arm or
`randomized
`
`studies can be
`used
`
`
`• Blinding
`preferred in
`
`comparative
`studies
`
`• Blinded review
`
`recommended
`
`
`
`• Rand iom zed
`
`studies essential
`• Blinding
`preferred
`• Blinded review
`
`recommended
`
`Objective
`Response
`
`Rate
`
`Surrogate for
`
`accelerated
`approval or
`
`regular approval*
`
`
`
`Complete
`Response
`
`
`
`Surrogate for
`
`accelerated
`approval or
`
`regular approval*
`
`
`agrro te for
`Su
`
`
`accelerated
`approval or
`
`regular approval*
`
`
`Progression-
`Free Survival
`(includes all
`deaths)
`or
`Time to
`Progression
`
`(deaths before
`
`progression
`
`censored)
`
`
`• Not a direct measure of
`benefit
`
`• Not a comprehensive
`measure of drug activity
`bset
`• Only a su
` of patients
`
`
`who benefit
`
`
`
`
`
`
`
`• Can be assessed in
`
`single-arm studies
`
`• Assessed earlier
`and in smaller
`studies compared
`with survival
`studies
`
`• Effect attributable
`to drug, not natural
`
`
`history
`
`
`
`
`• Not a direct measure of
`benefit in all cases
`• Not a comprehensive
`measure of drug activity
`• Small subset of patients
`
`with benefit
`
`
`
`• Can be assessed in
`
`single-arm studies
`
`• Durable complete
`responses can
`represent clinical
`benefit
`
`
`• Assessed earlier
`and in smaller
`studies compared
`with survival
`studies
`
`• Smaller sam e lp
`
`size and shorter
`follow-up
`
`necessary
`compared with
`survival studies
`
`• Measurement of
`stable disease
`
`included
`
`• Not affected by
`crossover or
`subsequent
`
`therapies
`
`• Generally based on
`
`objective and
`quantitative
`assessment
`*Adequacy as a surrogate endpoint for accelerated approval or regular approval is highly dependent upon other factors such as
`
`
`
`
`effect size, effect duration, and benefits of other available therapy. See text for details.
`
`
`
`A.
`
`Overall Survival
`
`Overall survival is defined as the time from randomization until death from any cause, and is
`measured in the intent-to-treat population. Survival is considered the most reliable cancer
`endpoint, and when studies can be conducted to adequately assess survival, it is usually the
`preferred endpoint. This endpoint is precise and easy to measure, documented by the date of
`
`5
`
`
`NPC02232929
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 8 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`
`death. Bias is not a factor in endpoint measurement. Survival improvement should be analyzed
`as a risk-benefit analysis to assess clinical benefit.
`
`Overall survival should be evaluated in randomized controlled studies. Data derived from
`
`historical trials are seldom reliable for time-dependent endpoints (e.g., overall survival, PFS).
`Apparent differences in outcome between historical controls and current treatment groups can
`arise from differences other than drug treatment, including patient selection, improved imaging
`techniques, or improved supportive care. Randomized studies minimize the effect of these
`differences by providing a direct outcome comparison. Demonstration of a statistically
`significant improvement in overall survival can be considered to be clinically significant if the
`toxicity profile is acceptable, and has often supported new drug approval.
`
`Difficulties in performing and analyzing survival studies include long follow-up periods in large
`trials and subsequent cancer therapy potentially confounding survival analysis.
`
`B.
`
`Endpoints Based on Tumor Assessments
`
`
`
`This section discusses several endpoints that are based on tumor assessments. These endpoints
`include DFS, ORR, TTP, PFS, and time-to-treatment failure (TTF). The collection and analysis
`of data on these time-dependent endpoints are based on indirect assessments, calculations, and
`estimates (e.g., tumor measurements). PFS data collection and analysis is supplemented by the
`tables shown in Appendix 3.
`
`Tumor-assessment endpoints selection should include two judgments. First, a determination of
`whether the endpoint will support either accelerated approval or regular approval should be
`ascertained. Second, the endpoint should be evaluated for the potential of bias or uncertainty in
`tumor endpoint assessments. Drug applications using studies that rely on tumor measurement-
`based endpoints as sole evidence of efficacy may need confirmatory evidence from a second
`trial. Accuracy in measuring tumors can differ among tumor settings. Tumor measurements
`used in response rate determinations can be imprecise in locations where there is a lack of
`
`demarcated margins (e.g., malignant mesothelioma, pancreatic cancer, brain tumors).
`
`When the primary study endpoint is based on tumor measurements (e.g., PFS or ORR), tumor
`endpoint assessments generally should be verified by central reviewers blinded to study
`treatments (see Appendix 4). This measure is especially important when the study itself is not
`blinded. It may be appropriate for the FDA to audit a sample of the scans to verify the central
`review process. Additional details regarding data collection are listed in Appendix 1.
`Centralized independent verification of tumor endpoint assessments (especially for PFS or DFS)
`may not be necessary when randomized trials are blinded (unless the adverse event profile would
`substantially unblind the trial in practice) or effect sizes are robust in large randomized trials
`
`where sensitivity analysis supports lack of observer bias (especially for DFS).
`
`
`
`1.
`
`Disease-Free Survival
`
`Generally, DFS is defined as the time from randomization until recurrence of tumor or death
`from any cause. The most frequent use of this endpoint is in the adjuvant setting after definitive
`
`
`
`6
`
`
`NPC02232930
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 9 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`
`surgery or radiotherapy. DFS also can be an important endpoint when a large percentage of
`patients achieve complete responses with chemotherapy. Although overall survival is a
`conventional endpoint for most adjuvant settings, DFS can be an important endpoint in situations
`where survival may be prolonged, making a survival endpoint impractical. DFS has been the
`primary basis of approval for adjuvant breast cancer hormonal therapy, adjuvant colon cancer,
`and adjuvant cytotoxic breast cancer therapy. Compared with standard cytotoxic therapies,
`hormonal therapies carry minimum side effects and thus a favorable risk-benefit relationship.
`DFS can be a surrogate for clinical benefit or it can provide direct evidence of clinical benefit.
`This determination is based on the magnitude of the effect, its risk-benefit relationship, and the
`disease setting. However, in disease settings where survival benefit has been already established,
`it is unlikely that DFS can be considered a clinical benefit. In December 2003, the ODAC
`consensus was DFS prolongation represented clinical benefit if the magnitude of this benefit
`outweighed the toxicity of the adjuvant treatment. In May 2004, the ODAC recommended that
`DFS be considered an acceptable endpoint for colon cancer drugs in the surgical adjuvant
`setting.7
`
`
`Important considerations in evaluating DFS as a potential endpoint include the estimated size of
`the treatment effect and proven benefits of standard therapies. The protocol should carefully
`delineate both the definition of DFS and the schedule for follow-up studies and visits.
`Unscheduled assessments can occur for many reasons and differences between study arms in the
`frequency, timing, or reason for unscheduled assessments can introduce bias. Bias can be
`minimized by blinding patients and investigators to the treatment assignments. The potential
`effects of bias due to unscheduled assessments can be evaluated by a comparative analysis of the
`total number of events over the follow-up period regardless of when the events occurred.
`
`The definition of DFS can be complicated, particularly when deaths are noted without prior
`tumor progression documentation. These events can be scored either as disease recurrences or as
`censored events. Although all methods for statistical analysis of deaths have some limitations,
`considering all deaths (deaths from all causes) as recurrences can minimize bias. DFS can be
`overestimated using this definition, especially in patients who die after a long period without
`observation. Bias can be introduced if the frequency of long-term follow-up visits is dissimilar
`between the study arms or if dropouts are not random because of toxicity. Some analyses count
`cancer-related deaths as DFS events and censor noncancer deaths. This method can introduce
`bias in the attribution of the cause of death. Furthermore, any method that censors patients,
`whether at death or at the last visit, assumes that the censored patients have the same risk of
`
`
`recurrence as noncensored patients.
`
`
`2.
`
`Objective Response Rate
`
`ORR is defined as the proportion of patients with tumor size reduction of a predefined amount
`and for a minimum time period. Response duration usually is measured from the time of initial
`response until documented tumor progression. Generally, the FDA has defined ORR as the sum
`of partial responses plus complete responses. When defined in this manner, ORR is a direct
`measure of drug antitumor activity, which can be evaluated in a single-arm study. Stable disease
`should not be a component of ORR. Stable disease can reflect the natural history of disease,
`
` 7 Transcripts are available at http://www.fda.gov/cder/drug/cancer_endpoints/default.htm.
`
`
`
`
`
`7
`
`
`NPC02232931
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 10 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`
`whereas tumor reduction is a direct therapeutic effect. Also, stable disease can be more
`accurately assessed by TTP or PFS analysis (see below). If available, standardized criteria
`should be used to ascertain response. A variety of response criteria have been considered
`appropriate (e.g., RECIST criteria) (Therasse and Arbuck et al., 2000, New Guidelines to
`Evaluate Response to Treatment in Solid Tumors, J Natl Cancer Inst, 92:205-16). The response
`criteria should be predefined in the protocol before the start of the study. The significance of
`ORR is assessed by its magnitude and duration, and the percentage of complete responses (no
`detectable evidence of tumor).
`
`3.
`
`Time to Progression and Progression-Free Survival
`
`
`
`
`TTP and PFS have served as primary endpoints for drug approval. TTP is defined as the time
`from randomization until objective tumor progression; TTP does not include deaths. PFS is
`defined as the time from randomization until objective tumor progression or death. The precise
`definition of tumor progression is important and should be carefully detailed in the protocol.
`
`a.
`
`TTP vs. PFS
`
`
`
`Compared with TTP, PFS is the preferred regulatory endpoint. PFS includes deaths and thus can
`
`be a better correlate to overall survival. In TTP analysis, deaths are censored, either at the time
`
`of death or at an earlier visit representing informative censoring (nonrandom pattern of loss from
`the study). PFS assumes patient deaths are randomly related to tumor progression. However, in
`situations where the majority of deaths are unrelated to cancer, TTP can be an acceptable
`
`endpoint.
`
`
`
`b.
`
`PFS as an endpoint to support drug approval
`
`Table 1 provides advantages and disadvantages of using PFS as an endpoint. PFS can reflect
`tumor growth and be assessed before the determination of a survival benefit. Its determination is
`not confounded by subsequent therapy. For a given sample size, the magnitude of effect on PFS
`can be larger than the effect on overall survival. However, the formal validation of PFS as a
`surrogate for survival for the many different malignancies that exist can be difficult. Data are
`usually insufficient to allow a robust evaluation of the correlation between effects on survival
`and PFS. Cancer trials are often small, and proven survival benefits of existing drugs are
`generally modest. The role of PFS as an endpoint to support licensing approval varies in
`different cancer settings. Whether an improvement in PFS represents a direct clinical benefit or
`a surrogate for clinical benefit depends on the magnitude of the effect and the risk-benefit of the
`new treatment compared to available therapies.
`
`
`
`c.
`
`PFS trial design issues
`
`The methodology for assessing, measuring, and analyzing PFS should be detailed in the protocol
`and statistical analysis plan (SAP). It is also important to carefully define tumor progression
`criteria in the protocol. There are no standard regulatory criteria for defining progression.
`Applicants have used a variety of different criteria, including the RECIST criteria. The broad
`outline presented in most published PFS criteria should be supplemented with additional details
`in the protocol and SAP. Visits and radiological assessments should be symmetric between the
`
`
`
`8
`
`
`NPC02232932
`
`NOVARTIS EXHIBIT 2150
`Par v Novartis, IPR 2016-00084
`Page 11 of 22
`
`

`
`Contains Nonbinding Recommendations
`
`
`
`• The study design
`• The definition of progression
`• The data to be recorded on the case report form (CRF)
`• The SAP
`• The methodology for handling missing data and censoring methods
`• The operating procedures of an independent endpoint review committee (IRC), if
`
`applicable (see Appendix 4)
`
`
`d.
`
`Analysis of PFS
`
`
`two study arms to prevent systematic bias. When possible, studies should be blinded. Blinding
`is particularly important when patient or investigator assessments are included as components of
`the progression endpoint. At a minimum, the assessments should be subjected to a blinded
`independent adjudication team, generally consisting of radiologists and clinicians. The FDA and
`the applicant should agree prospectively on the following items:
`
`
`
`
`Missing data can complicate analysis of PFS. The protocol should define a

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket